WO2006042848A2 - Conjugues d'hormones de croissance - Google Patents

Conjugues d'hormones de croissance Download PDF

Info

Publication number
WO2006042848A2
WO2006042848A2 PCT/EP2005/055336 EP2005055336W WO2006042848A2 WO 2006042848 A2 WO2006042848 A2 WO 2006042848A2 EP 2005055336 W EP2005055336 W EP 2005055336W WO 2006042848 A2 WO2006042848 A2 WO 2006042848A2
Authority
WO
WIPO (PCT)
Prior art keywords
meo
compound according
tetrazolyl
compound
formula
Prior art date
Application number
PCT/EP2005/055336
Other languages
English (en)
Other versions
WO2006042848A9 (fr
WO2006042848A3 (fr
Inventor
Florenzio Zaragoza DÖRWALD
Lars Fogh Iversen
Nils Langeland Johansen
Original Assignee
Novo Nordisk A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novo Nordisk A/S filed Critical Novo Nordisk A/S
Priority to JP2007537265A priority Critical patent/JP2008516621A/ja
Priority to US11/577,238 priority patent/US20080182783A1/en
Priority to EP05797317A priority patent/EP1805216A2/fr
Publication of WO2006042848A2 publication Critical patent/WO2006042848A2/fr
Publication of WO2006042848A3 publication Critical patent/WO2006042848A3/fr
Publication of WO2006042848A9 publication Critical patent/WO2006042848A9/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormone [GH], i.e. somatotropin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to conjugates of growth hormones with improved pharmacological properties, and to the use of said hormones in therapy.
  • regioselective conjugation It is often desired or even required to conjugate at specific site(s), and this is referred to as regioselective conjugation.
  • Regioselective acylation of growth hormones such as e.g. human growth hormone (hGH) is, however, difficult, because this protein contains nine lysine residues of similar reactivity, and mixtures of products usually results. The single components of these mixtures are difficult to isolate, and will usually be obtained in low yield and purity only.
  • Gaertner et al in Bioconjugate Chem., 7, 38-44, 1996 disclose a method for conjugating PEG to IL-8, G-CFS and IKL-1 ra by generating an aldehyde at the N-terminus, followed by reaction with an alkoxyamine functionalised PEG.
  • the aldehyde is generated at the N-terminus either by oxidation with periodate if the N-terminal amino acid residue is serine, or by metal catalysed transamination if the N-terminal amino acid residue is different from serine.
  • N-terminal serine extended human growth hormone, Ser-hGH is disclosed as SEQ
  • human growth hormone may be reductively alkylated with aldehydes selectively at the N-terminal amino group (US 20040127417).
  • the present invention relates to growth hormone compounds (GH) selectively conjugated at the N-terminal to improve pharmacological properties compared to the parent growth hormone compound. Accordingly, in one embodiment the invention provides compounds of formula (I)
  • GH represent a radical derived from a growth hormone compound by removal of one hydrogen atom from the N-terminal amino group
  • X represents oxygen or two hydrogen atoms
  • Z represents a bond, alkylene, arylene, heteroarylene, -CH 2 O-(CH 2 )i-io-, -CH 2 -O-(C 6 H 4 )-, or combinations thereof;
  • Y represents a radical selected from
  • R 2 , R 3 , R 5 and R 6 independently represent
  • R 2 and R 3 does not represent H or Ci -6 alkyl ;
  • E represents a bond or a diradical selected from
  • the invention relates to methods of preparing compounds of formula I, said method comprising the steps of
  • A represents an aldehyde or a ketone moiety
  • the invention provides a compound according to formula IV or V,
  • the invention provides the use of a compound according to formula (II) or formula (III) in the preparation of conjugated GH with improved pharmacological properties compared to the parent GH. In one embodiment, the invention provides the use of a compound according to formula (IV) or formula (V) in the preparation of conjugated GH with improved pharmacological properties compared to the parent GH.
  • the invention relates to a nucleic acid construct comprising a nucleic acid sequence encoding a compound according to formula V, provided that said construct does not encode Ser-hGH, to a vector comprising said construct, and to a host cell comprising said vector.
  • the invention provides compounds according to formula (I) for use in therapy.
  • the invention provides a pharmaceutical composition comprising a compound according formula (I).
  • the invention provides a method of treating diseases benefiting from an increase in the level of circulating growth hormone, the method comprising the administration of a therapeutically effective amount of a compound according to formula (I) to a patient in need thereof.
  • the invention relates to the use of a compound according to formula (I) in the manufacture of a medicament for the treatment of a disease benefiting from an increase in the level of circulating growth hormone.
  • growth hormone is intended to indicate a protein which exhibits growth hormone activity as determined in assay I herein.
  • a protein which exhibits an activity above 20%, such as above 40%, such as above 60%, such as above 80% of that of hGH in said assay is defined as a growth hormone compound.
  • a protein is intended to indicate a sequence of two or more amino acids joined by peptide bonds, wherein said amino acids may be natural or un-natural. It is to be understood that the term is also intended to include proteins which have been derivatized, e.g. by the attachment of lipophilic groups, PEG or prosthetic groups.
  • alkyl is intended to indicate a straight, branched and/or cyclic saturated monovalent hydrocarbon radical having from one to six carbon atoms, also denoted as Ci- 6 -alkyl.
  • Ci- 6 -alkyl groups include e.g. methyl, ethyl, n-propyl, isopropyl, 8
  • alkylene indicates the corresponding bi-radical.
  • aryl as used herein is intended to indicate carbocyclic aromatic ring systems comprising one or more rings, such as phenyl, biphenylyl, naphthyl, anthracenyl, phenanthrenyl, fluorenyl, indenyl, pentalenyl and azulenyl.
  • Aryl is also intended to include the partially hydrogenated derivatives of the multi-ring carbocyclic systems enumerated above, wherein at least one ring is aromatic. Examples of such partially hydrogenated derivatives include 1 ,2,3,4-tetrahydronaphthyl and 1 ,4-dihydronaphthyl.
  • arylene is intended to indicate the corresponding bi-radical, and examples include 1 ,2-phenylene, 1 ,3-phenylene, 1 ,4-phenylene, 1 ,2-naphthylene, 1 ,4-naphthylene, 4,4'-biphenylene, 4,4"-terphenylene and 4,4'"-quaterphenylene.
  • heteroaryl as used herein is intended to indicate radicals of heterocyclic aromatic ring systems containing one or more heteroatoms selected from nitrogen, oxygen and sulphur, such as furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, isoxazolyl, isothiazolyl, 1 ,2,3-triazolyl, 1 ,2,4-triazolyl, pyranyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1 ,2,3-triazinyl, 1 ,2,4-triazinyl, 1 ,3,5- triazinyl, 1 ,2,3-oxadiazolyl, 1 ,2,4-oxadiazolyl, 1 ,2,5- oxadiazolyl, 1 ,3,4-oxadiazolyl, 1 ,2,3-thiadiazolyl, 1
  • the term is also intended to include partially hydrogenated derivatives of the multi-ring heterocyclic systems enumerated above, provided at least one ring comprising a hetero atom is aromatic.
  • partially hydrogenated derivatives include 2,3-dihydrobenzofuranyl, pyrrolinyl, pyrazolinyl, indolinyl, oxazolidinyl, oxazolinyl and oxazepinyl.
  • heteroarylene is intended to indicate the corresponding bi-radical, and examples include 1 ,2,4-pyrazol-2,5-diyl, imidazol-1 ,2-diyl, thiazol-2,4-diyl, (4-phenylimidazole)-4,1 '-diyl and (3,5-diphenyl-1 ,2,4-oxadiazole)-4,4"-diyl.
  • cibacronyl means the radical sketched below, or any salt or solvate of the same:
  • solvate is a complex of defined stoichiometry formed by a solute and a solvent.
  • Solvents may be, by way of example, water, ethanol, or acetic acid.
  • dipolar solvent refers to a solvent with a dielectric constant larger than 6.0.
  • Peg means a polydisperse or monodisperse diradical of the structure
  • n is an integer larger than 1 , and its molecular weight is between approximately 100 and approximately 1 ,000,000 Da.
  • mPEG or "mPeg” means a polydisperse or monodisperse radical of the structure
  • m is an integer larger than 1 .
  • an mPEG wherein m is 90 has a molecular weight of 3991 Da, i.e. approximately 4kDa.
  • an mPEG with an average molecular weight of 20 kDa has an average m of 454. Due to the process for producing mPEG these molecules often have a distribution of molecular weights. This distribution is described by the polydispersity index.
  • mPEG with a molecular weight of 20 kDa may also be referred to as MeO-(CH 2 CH 2 O) 40 O-SOo
  • mPEG with a molecular weight of 30 kDa may also be referred to as MeO-(CH 2 CH 2 0) 6 oo- 7 oo
  • mPEG with a molecular weight of 40 kDa may also be referred to as MeO-(CH 2 CH 2 0) 85 o- 9 5o-
  • the heavier mPEG chains may be difficult to prepare as a single chain molecule, and they are thus made as branched mPEG.
  • mPEG with a molecular weight of 40 kDa may be achieved with as a branched mPEG comprising to arms of 20 kDa each.
  • polydispersity index means the ratio between the weight average molecular weight and the number average molecular weight, as known in the art of 10
  • the polydispersity index is a number which is greater than or equal to one, and it may be estimated from Gel Permeation Chromatographic data. When the polydispersity index is 1 , the product is monodisperse and is thus made up of compounds with a single molecular weight. When the polydispersity index is greater than 1 it is a measure of the polydispersity of that polymer, i.e. how broad the distribution of polymers with different molecular weights is.
  • mPEG20000 in formulas, compound names or in molecular structures indicates an mPEG residue wherein mPEG is polydisperse and has a molecular weight of approximately 20 kDa.
  • the polydispersity index typically increases with the molecular weight of the PEG or mPEG.
  • a polydisperisty index below 1 .06, such as below 1 .05, such as below 1.04, such as below 1.03, such as between 1 .02 and 1.03.
  • prodrug includes biohydrolyzable amides and biohydrolyzable esters and also encompasses a) compounds in which the biohydrolyzable functionality in such a prodrug is encompassed in the compound according to the present invention, and b) compounds which may be oxidized or reduced biologically at a given functional group to yield drug substances according to the present invention.
  • these functional groups include 1 ,4-dihydropyridine, N-alkylcarbonyl-1 ,4-dihydropyridine, 1 ,4- cyclohexadiene, tert-butyl, and the like.
  • biohydrolyzable ester is an ester of a drug substance (in casu, a compound according to the invention) which either a) does not interfere with the biological activity of the parent substance but confers on that substance advantageous properties in vivo such as duration of action, onset of action, and the like, or b) is biologically inactive but is readily converted in vivo by the subject to the biologically active principle.
  • the advantage is, for example increased solubility or that the biohydrolyzable ester is orally absorbed from the gut and is transformed to a compound according to the present invention 11
  • lower alkyl esters e.g., C 1 -C 4
  • lower acyloxyalkyl esters lower alkoxyacyloxyalkyl esters
  • alkoxyacyloxy esters alkyl acylamino alkyl esters
  • choline esters e.g., choline esters
  • biohydrolyzable amide is an amide of a drug substance (in casu, a compound according to the present invention) which either a) does not interfere with the biological activity of the parent substance but confers on that substance advantageous properties in vivo such as duration of action, onset of action, and the like, or b) is biologically inactive but is readily converted in vivo by the subject to the biologically active principle.
  • the advantage is, for example increased solubility or that the biohydrolyzable amide is orally absorbed from the gut and is transformed to a compound according to the present invention in plasma.
  • Many examples of such are known in the art and include by way of example lower alkyl amides, ⁇ -amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides.
  • salts are intended to indicate salts which are not harmful to the patient.
  • Such salts include pharmaceutically acceptable acid addition salts, pharmaceutically acceptable metal salts, ammonium and alkylated ammonium salts.
  • Acid addition salts include salts of inorganic acids as well as organic acids. Representative examples of suitable inorganic acids include hydrochloric, hydrobromic, hydroiodic, phosphoric, sulfuric, nitric acids and the like.
  • suitable organic acids include formic, acetic, trichloroacetic, trifluoroacetic, propionic, benzoic, cinnamic, citric, fumaric, glycolic, lactic, maleic, malic, malonic, mandelic, oxalic, picric, pyruvic, salicylic, succinic, methanesulfonic, ethanesulfonic, tartaric, ascorbic, pamoic, bismethylene salicylic, ethanedisulfonic, gluconic, citraconic, aspartic, stearic, palmitic, EDTA, glycolic, p-aminobenzoic, glutamic, benzenesulfonic, p-toluenesulfonic acids and the like.
  • compositions include the pharmaceutically acceptable salts listed in J. Pharm. Sci. 1977, 66, 2, which is incorporated herein by reference.
  • metal salts include lithium, sodium, potassium, magnesium salts and the like.
  • ammonium and alkylated ammonium salts include ammonium, methylammonium, dimethylammonium, trimethylammonium, ethylammonium, hydroxyethylammonium, diethylammonium, butylammonium, tetramethylammonium salts and the like.
  • a “therapeutically effective amount” of a compound as used herein means an amount sufficient to cure, alleviate or partially arrest the clinical manifestations of a given disease and its complications. An amount adequate to accomplish this is defined as “therapeutically effective amount”. Effective amounts for each purpose will depend on e.g. 12
  • treatment means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder.
  • the term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or complications, to delay the progression of the disease, disorder or condition, to alleviate or relief the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complications.
  • the patient to be treated is preferably a mammal, in particular a human being, but it may also include animals, such as dogs, cats, cows, sheep and pigs.
  • the present invention relates to GH to which a group has been conjugated at the N-terminal, i.e. to compounds according to formula (I)
  • Said compounds have improved pharmacological properties compared to the corresponding un-conjugated GH, also referred to as the parent GH.
  • pharmacological properties include functional in vivo half-life, immunogenicity, renal filtration, protease protection and albumin binding.
  • the term "functional in vivo half-life” is used in its normal meaning, i.e., the time at which 50% of the biological activity of the GH or conjugated GH are still present in the body/target organ, or the time at which the activity of the GH or GH conjugate is 50% of its initial value.
  • in vivo plasma half- life may be determined, i.e., the time at which 50% of the GH or GH conjugate circulate in the plasma or bloodstream prior to being cleared. Determination of plasma half-life is often more simple than determining functional half-life and the magnitude of plasma half-life is usually a good indication of the magnitude of functional in vivo half-life.
  • plasma half-life include serum half-life, circulating half-life, circulatory half-life, serum clearance, plasma clearance, and clearance half-life.
  • the term "increased" as used in connection with the functional in vivo half-life or plasma half-life is used to indicate that the relevant half-life of the GH conjugate is statistically significantly increased relative to that of the parent GH, as determined under comparable conditions.
  • the relevant half-life may be increased by at least about 25%, such as by at lest about 50%, e.g., by at least about 100%, 150%, 200%, 250%, or 500%.
  • the compounds of the present invention exhibit an increase in half-life of at least about 5 h, preferably at least about 24 h, more preferably at least about 72 h, and most preferably at least about 7 days, relative to the half-life of the parent GH.
  • Measurement of in vivo plasma half-life can be carried out in a number of ways as described in the literature.
  • An increase in in vivo plasma half-life may be quantified as a decrease in clearance (CL) or as an increase in mean residence time (MRT).
  • Conjugated GH of the present invention for which the CL is decreased to less than 70%, such as less than 50%, such than less than 20%, such than less than 10% of the CL of the parent GH as determined in a suitable assay is said to have an increased in vivo plasma half-life.
  • Conjugated GH of the present invention for which MRT is increased to more than 130%, such as more than 150%, such as more than 200%, such as more than 500% of the MRT of the parent GH in a suitable assay is said to have an increased in vivo plasma half-life. Clearance and mean residence time can be assessed in standard pharmacokinetic studies using suitable test animals. It is within the capabilities of a person skilled in the art to choose a suitable test animal for a given protein. Tests in human, of course, represent the ultimate test. Suitable text animals include normal, Sprague-Dawley male rats, mice and cynomolgus monkeys.
  • mice and rats are in injected in a single subcutaneous bolus, while monkeys may be injected in a single subcutaneous bolus or in a single iv dose.
  • the amount injected depends on the test animal.
  • blood samples are taken over a period of one to five days as appropriate for the assessment of CL and MRT. The blood samples are conveniently analysed by ELISA techniques.
  • Immunogenicity refers to the ability of the compound, when administered to a human, to elicit a deleterious immune response, whether humoral, cellular, or both. In any human sub-population, there may exist individuals who exhibit sensitivity to particular administered proteins. Immunogenicity may be measured by quantifying the presence of growth hormone antibodies and/or growth hormone responsive T-cells in a sensitive individual, using conventional methods known in the art. In one embodiment, the conjugated GH of the present invention exhibit a decrease in 14
  • immunogenicity in a sensitive individual of at least about 10%, preferably at least about 25%, more preferably at least about 40% and most preferably at least about 50%, relative to the immunogenicity for that individual of the parent GH.
  • protease protection or “protease protected” as used herein is intended to indicate that the conjugated GH of the present invention is more resistant to the plasma peptidase or proteases than is the parent GH.
  • Protease and peptidase enzymes present in plasma are known to be involved in the degradation of circulating proteins, such as e.g. circulating peptide hormones, such as growth hormone.
  • DPPIV dipeptidyl aminopeptidase IV
  • Proteins and their degradation products may be monitored by their absorbance at 220 nm (peptide bonds) or 280 nm (aromatic amino acids), and are quantified by integration of their peak areas related to those of standards.
  • the rate of hydrolysis of a protein by dipeptidyl aminopeptidase IV is estimated at incubation times which result in less than 10% of the peptide being hydrolysed.
  • the rate of hydrolysis of the GH conjugate is less than 70%, such as less than 40%, such as less than 10% of that of the parent GH.
  • Serum albumin The most abundant protein component in circulating blood of mammalian species is serum albumin, which is normally present at a concentration of approximately 3 to 4.5 grams per 100 milliters of whole blood.
  • Serum albumin is a blood protein of approximately 70,000 daltons which has several important functions in the circulatory system. It functions as a transporter of a variety of organic molecules found in the blood, as the main transporter of various metabolites such as fatty acids and bilirubin through the blood, and, owing to its abundance, as an osmotic regulator of the circulating blood.
  • Serum albumin has a half-life of more than one week, and one approach to increasing the plasma half-life of proteins has been to conjugate to the protein a group that binds to serum albumin.
  • GH is human growth hormone (hGH) which has an amino acid sequence as set forth in SEQ ID NO 1 .
  • GH is a variant of hGH, wherein a variant is understood to be the compound obtained by substituting one or more amino acid residues in the hGH sequence with another natural or unnatural amino acid; and/or by adding one or more natural or unnatural amino acids to the hGH sequence; and/or by deleting one or more amino acid residue from the hGH sequence, wherein any of these steps may optionally be followed by further derivatization of one or more amino acid residue.
  • substitutions are conservative in the sense that one amino acid residue is substituted by another amino acid residue from the same group, i.e. by another amino acid residue with similar properties.
  • Amino acids may conveniently be divided in the following groups based on their properties: Basic amino acids (such as arginine, lysine, histidine), acidic amino acids (such as glutamic acid and aspartic acid), polar amino acids (such as glutamine, cysteine and asparagine), hydrophobic amino acids (such as leucine, isoleucine, proline, methionine and valine), aromatic amino acids (such as phenylalanine, tryptophan, tyrosine) and small amino acids (such as glycine, alanine, serine and threonine.).
  • Basic amino acids such as arginine, lysine, histidine
  • acidic amino acids such as glutamic acid and aspartic acid
  • polar amino acids such as glutamine, cysteine and asparagine
  • hydrophobic amino acids such as leucine, isoleucine, proline, methionine and valine
  • aromatic amino acids such as phenylalanine, try
  • GH has at least 80%, such as at least 85%, such as at least 90%, such as at least 95% identity with hGH.
  • said identities to hGH is coupled to at least 20%, such as at least 40%, such as at least 60%, such as at least 80% of the growth hormone activity of hGH as determined in assay I herein.
  • identity refers to a relationship between the sequences of two or more proteins, as determined by comparing the sequences.
  • identity also means the degree of sequence relatedness between proteins, as determined by the number of matches between strings of two or more amino acid residues.
  • Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., "algorithms").
  • Identity of related proteins can be readily calculated by known methods. Such methods include, but are not limited to, those described in Computational
  • Preferred methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity are described in publicly available computer programs. Preferred computer program methods to determine identity between two sequences include the GCG program package, including GAP (Devereux et al., Nucl. Acid. Res., 12:387 (1984); Genetics Computer Group, University of Wisconsin, Madison, Wis.), BLASTP, BLASTN, and FASTA (Altschul et al., J. MoI. Biol., 215:403-410 (1990)). The BLASTX program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Altschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Altschul et al., supra). The well known Smith Waterman algorithm may also be used to determine identity.
  • NCBI National Center for Biotechnology Information
  • a gap opening penalty (which is calculated as 3.times. the average diagonal; the "average diagonal” is the average of the diagonal of the comparison matrix being used; the “diagonal” is the score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually ⁇ fraction (1/10) ⁇ times the gap opening penalty), as well as a comparison matrix such as PAM 250 or BLOSUM 62 are used in conjunction with the algorithm.
  • a standard comparison matrix (see Dayhoff et al., Atlas of Protein Sequence and Structure, vol. 5, supp.3 (1978) for the PAM 250 comparison matrix; Henikoff et al., Proc. Natl. Acad. Sci USA, 89:10915-10919 (1992) for the BLOSUM 62 comparison matrix) is also used by the algorithm.
  • Preferred parameters for a protein sequence comparison include the following: Algorithm: Needleman et al., J. MoI. Biol, 48:443-453 (1970); Comparison matrix: BLOSUM 62 from Henikoff et al., Proc. Natl. Acad. Sci. USA, 89:10915-10919 (1992); Gap Penalty: 12, Gap Length Penalty: 4, Threshold of Similarity: 0.
  • the GAP program is useful with the above parameters.
  • the aforementioned parameters are the default parameters for protein comparisons (along with no penalty for end gaps) using the GAP algorithm.
  • GH is hGH extended with up to 100 amino acid residues at the N-terminal.
  • said extension is up to 50, such as up to 40, such as up to 20, such as up to 10, such as up to 5, such as 1 , 2 or 3 amino acid residues. 17
  • X represents two hydrogens in which case compounds of formula (I) take the form of formula (T)
  • R 9 represents hydrogen.
  • R 1 may represent MeO-(CH 2 CH 2 0) 40 o- 5 oo-E-, MeO-(CH 2 CH 2 0) 60 o- 7 oo-E- or MeO-
  • Z represents -CH 2 -O-(CH 2 ) 1 . 5 or -CH-O- (C 6 H 4 )-.
  • Z may represent -CH 2 -O-(CH 2 ) 3 , -CH 2 -O-(CH 2 ) 4 , -CH 2 -O-(CH 2 ) 5 , -CH 2 -O-(1 ,4-C 6 H 4 )- or -CH 2 -O-(1 ,3-C 6 H 4 )-.
  • Y represents 18
  • R 9 represents hydrogen
  • R1 represents
  • Y represents
  • R 6 and R 9 represent hydrogen, R 7 and R 8 independently represent hydrogen or methyl, and R 5 represents 21
  • R 6 and R 9 represent hydrogen
  • R 7 and R 8 independently represent hydrogen or methyl
  • R 5 represents
  • R 1 represents 23
  • the GH in the above compounds is hGH.
  • the invention provides the methods and compounds described herein, wherein each instance of "mPEG” is replaced by an alkoxy- PEG or "aPEG” compound of the formula
  • Each R1 can be any suitable C 1 - 1 0 alkyl group, branched or (for C3- 1 0) unbranched, including, but not limited to, methyl, ethyl, and propyl, and butyl. 30
  • the invention relates a method for preparing compounds of formula (I) or (I 1 ), the method comprising the steps of
  • A represents an aldehyde or ketone moiety
  • Q is selected from
  • R 9 represents H, d- 6 alkyl, aryl, or heteroaryl, and compounds of formula (III 1 ) takes the form of formula (III)
  • DMF is one example of a dipolar solvent.
  • the invention relates to a method for preparing compounds according to formula (I) or (I"), the method comprising the steps of
  • the oxidation in step (a') is brought about by mixing a buffered solution of GH (pH 7-9) with a solution of methionine (30-300 equivalents), and adding a solution of periodate, such as NaIO 4 (5-20 equivalents).
  • GH pH 7-9
  • methionine a solution of methionine
  • periodate such as NaIO 4
  • Particular examples of compounds of formula (IV) or (V) include Ser-hGH, and
  • the invention relates to methods of improving the pharmacological properties of GH, the method comprising preparing a conjugated GH as described in the above methods.
  • improving the pharmacological properties is intended to indicate an increase in the functional in vivo half-life, the plasma in vivo half-life, the mean residence time, or an decrease in the clearance.
  • the invention provides a method for the treatment of growth hormone deficiency (GHD); Turner Syndrome; Prader-Willi syndrome (PWS); Noonan syndrome; Down syndrome; chronic renal disease, juvenile rheumatoid arthritis; cystic fibrosis, HIV-infection in children receiving HAART treatment (HIV/HALS children); short children born short for gestational age (SGA); short stature in children born with very low birth weight (VLBW) but SGA; skeletal dysplasia; hypochondroplasia; achondroplasia; idiopathic short stature (ISS); GHD in adults; fractures in or of long bones, such as tibia, fibula, femur, humerus, radius, ulna, clavicula, matacarpea, matatarsea, and digit; fractures
  • APCD chronic dialysis
  • malnutritional associated cardiovascular disease in APCD reversal of cachexia in APCD; cancer in APCD; chronic abstractive pulmonal disease in APCD; HIV in APCD; elderly with APCD; chronic liver disease in APCD, fatigue syndrome in APCD; Crohn's disease; impaired liver function; males with HIV infections; short bowel syndrome; central obesity; HIV-associated lipodystrophy syndrome (HALS); male infertility; patients after major elective surgery, alcohol/drug detoxification or neurological trauma; aging; frail elderly; osteo-arthritis; traumatically damaged cartilage; erectile dysfunction; fibromyalgia; memory disorders; depression; traumatic brain injury; traumatic spinal cord injury; subarachnoid haemorr
  • the invention provides a method for the acceleration of the healing of muscle tissue, nervous tissue or wounds; the acceleration or improvement of blood flow to damaged tissue; or the decrease of infection rate in damaged tissue, the method comprising administration to a patient in need thereof an effective amount of a therapeutically effective amount of a compound of formula I.
  • the invention relates to the use of compounds according to formula I in the manufacture of diseases benefiting from an increase in the growth hormone plasma level, such as the disease mentioned above.
  • a typical parenteral dose is in the range of 10 ⁇ 9 mg/kg to about 100 mg/kg body weight per administration.
  • Typical administration doses are from about 0.0000001 to about 10 mg/kg body weight per administration.
  • the exact dose will depend on e.g. indication, medicament, frequency and mode of administration, the sex, age and general condition of the subject to be treated, the nature and the severity of the disease or condition to be treated, the desired effect of the treatment and other factors evident to the person skilled in the art.
  • Typical dosing frequencies are twice daily, once daily, bi-daily, twice weekly, once weekly or with even longer dosing intervals. Due to the prolonged half-lifes of the fusion proteins of the present invention, a dosing regime with long dosing intervals, such as twice weekly, once weekly or with even longer dosing intervals is a particular embodiment of the invention.
  • the GH and in particular Ser-GH may be prepared in a number of different ways, such as e.g. synthesis using standard protein synthesis techniques. In a particular embodiment, however, the GH or Ser-GH is expressed in a suitable host after incorporation of a suitable nucleic acid construct into said host.
  • nucleic acid construct is intended to indicate any nucleic acid molecule of cDNA, genomic DNA, synthetic DNA or RNA origin.
  • construct is intended to indicate a nucleic acid segment which may be single- or double-stranded, and 34
  • the construct may be based on a complete or partial nucleotide sequence encoding a protein of interest.
  • the construct may optionally contain other nucleic acid segments.
  • the nucleic acid construct of the invention encoding the protein of the invention may suitably be of genomic or cDNA origin, for instance obtained by preparing a genomic or cDNA library and screening for DNA sequences coding for all or part of the protein by hybridization using synthetic oligonucleotide probes in accordance with standard techniques (cf. J. Sambrook, E. F. Fritsch, and T. Maniatus, 1989, Molecular Cloning, A Laboratory Manual, 2d edition, Cold Spring Harbor, New York).
  • the DNA sequence encoding the GH is preferably of human origin, i.e. derived from a human genomic DNA or cDNA library.
  • the DNA sequence may be of human origin, e.g. cDNA from a particular human organ or cell type or a gene derived from human genomic DNA.
  • the nucleic acid construct of the invention encoding the GH or Ser-GH may also be prepared synthetically by established standard methods, e.g. the phosphoamidite method described by Beaucage and Caruthers, Tetrahedron Letters 22 (1981 ), 1859 - 1869, or the method described by Matthes et al., EMBO Journal 3 (1984), 801 - 805.
  • phosphoamidite method oligonucleotides are synthesized, e.g. in an automatic DNA synthesizer, purified, annealed, ligated and cloned in suitable vectors.
  • nucleic acid construct may be of mixed synthetic and genomic, mixed synthetic and cDNA or mixed genomic and cDNA origin prepared by ligating fragments of synthetic, genomic or cDNA origin (as appropriate), the fragments corresponding to various parts of the entire nucleic acid construct, in accordance with standard techniques.
  • the nucleic acid construct may also be prepared by polymerase chain reaction using specific primers, for instance as described in US 4,683,202 or Saiki et al., Science 239 (1988), 487 - 491.
  • the nucleic acid construct is a DNA construct which term will be used exclusively in the following.
  • the present invention relates to a recombinant vector comprising a DNA construct of the invention.
  • the recombinant vector into which the DNA construct of the invention is inserted may be any vector which may conveniently be subjected to recombinant DNA procedures, and the choice of vector will often depend on the host cell into which it is to be introduced.
  • the vector may be an autonomously replicating vector, i.e. a vector which exists as an extrachromosomal entity, the replication of which is independent of chromosomal replication, e.g. a plasmid.
  • the vector may be one 35
  • the vector is preferably an expression vector in which the DNA sequence encoding the protein of the invention is operably linked to additional segments required for transcription of the DNA.
  • the expression vector is derived from plasmid or viral DNA, or may contain elements of both.
  • operably linked indicates that the segments are arranged so that they function in concert for their intended purposes, e.g. transcription initiates in a promoter and proceeds through the DNA sequence coding for the protein.
  • the promoter may be any DNA sequence which shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • Suitable promoters for directing the transcription of the DNA encoding the GH in mammalian cells are the SV40 promoter (Subramani et al., MoI. Cell Biol. 1 (1981 ), 854 -864), the MT- 1 (metallothionein gene) promoter (Palmiter et al., Science 222 (1983), 809 - 814) or the adenovirus 2 major late promoter.
  • a suitable promoter for use in insect cells is the polyhedrin promoter (US 4,745,051 ; Vasuvedan et al., FEBS Lett. 31 1. (1992) 7 - 11 ), the P10 promoter (J. M. Vlak et al., J. Gen. Virology 69, 1988, pp. 765-776), the Autographa californica polyhedrosis virus basic protein promoter (EP 397 485), the baculovirus immediate early gene 1 promoter (US 5,155,037; US 5,162,222), or the baculovirus 39K delayed-early gene promoter (US 5,155,037; US 5,162,222).
  • the polyhedrin promoter US 4,745,051 ; Vasuvedan et al., FEBS Lett. 31 1. (1992) 7 - 11
  • the P10 promoter J. M. Vlak et al., J. Gen. Virology 69, 1988, pp. 765-776
  • promoters for use in yeast host cells include promoters from yeast glycolytic genes (Hitzeman et al., J. Biol. Chem. 255 (1980). 12073 - 12080; Alber and Kawasaki, J. MoI. Appl. Gen. 1 (1982), 419 - 434) or alcohol dehydrogenase genes (Young et al., in Genetic Engineering of Microorganisms for Chemicals (Hollaender et al, eds.), Plenum Press, New York, 1982), or the TPH (US 4,599,31 1 ) or ADH2-4c (Russell et al., Nature 304 (1983), 652 - 654) promoters.
  • suitable promoters for use in filamentous fungus host cells are, for instance, the ADH3 promoter (McKnight et al., The EMBO J. 4 (1985), 2093 - 2099) or the tpjA promoter.
  • suitable promoters are those derived from the gene encoding A. oryzae TAKA amylase, Rhizomucor miehei aspartic proteinase, A. niger neutral ⁇ - amylase, A. niger acid stable ⁇ -amylase, A. niger or A. awamori glucoamylase (gluA), Rhizo ⁇ mucor miehei lipase, A. oryzae alkaline protease, A. oryzae triose phosphate isomerase or A. nidulans acetamidase.
  • Preferred are the TAKA-amylase and gluA promoters. 36
  • suitable promoters for use in bacterial host cells include the promoter of the Bacillus stearothermophilus maltogenic amylase gene, the Bacillus licheniformis alpha- amylase gene, the Bacillus amyloliquefaciens BAN amylase gene, the Bacillus subtilis alkaline protease gen, or the Bacillus pumilus xylosidase gene, or by the phage Lambda P R or P L promoters or the E. coli lac, trp_ or tac promoters.
  • the DNA sequence encoding GH or Ser-GH may also, if necessary, be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter et al., o ⁇ . cit) or (for fungal hosts) the TPH (Alber and Kawasaki, p_
  • the vector may further comprise elements such as polyadenylation signals (e.g. from SV40 or the adenovirus 5 EIb region), transcriptional enhancer sequences (e.g. the SV40 enhancer) and translational enhancer sequences (e.g. the ones encoding adenovirus VA RNAs).
  • the recombinant vector of the invention may further comprise a DNA sequence enabling the vector to replicate in the host cell in question.
  • a DNA sequence enabling the vector to replicate in the host cell in question.
  • An example of such a sequence is the SV40 origin of replication.
  • suitable sequences enabling the vector to replicate are the yeast plasmid 2 ⁇ replication genes REP 1 -3 and origin of replication.
  • sequences enabling the vector to replicate are DNA polymerase III complex encoding genes and origin of replication.
  • the vector may also comprise a selectable marker, e.g. a gene the product of which complements a defect in the host cell, such as the gene coding for dihydrofolate reductase (DHFR) or the Schizosaccharomyces pombe TP ⁇ gene (described by P. R. Russell, Gene 40, 1985, pp. 125-130), or one which confers resistance to a drug, e.g. ampicillin, kanamycin, tetracyclin, chloramphenicol, neomycin, hygromycin or methotrexate.
  • selectable markers include amdS, pyrG, arqB, niaD and sC.
  • a secretory signal sequence (also known as a leader sequence, prepro sequence or pre sequence) may be provided in the recombinant vector.
  • the secretory signal sequence is joined to the DNA sequence encoding the protein in the correct reading frame.
  • Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the protein.
  • the secretory signal sequence may be that which is normally associated with the GH or may be from a gene encoding another secreted protein.
  • the secretory signal sequence may encode any signal peptide which ensures efficient direction of the expressed protein into the secretory pathway of the cell.
  • the signal peptide may be naturally occurring signal peptide, or a 37
  • Suitable signal peptides have been found to be the ⁇ -factor signal peptide (cf. US 4,870,008), the signal peptide of mouse salivary amylase (cf. O. Hagenbuchle et al., Nature 289, 1981 , pp. 643-646), a modified carboxypeptidase signal peptide (cf. L.A. VaIIs et al., Cell 48, 1987, pp. 887-897), the yeast BAR1 signal peptide (cf. WO 87/02670), or the yeast aspartic protease 3 (YAP3) signal peptide (cf. M. Egel-Mitani et al., Yeast 6, 1990, pp. 127-137).
  • a sequence encoding a leader peptide may also be inserted downstream of the signal sequence and uptream of the DNA sequence encoding the protein.
  • the function of the leader peptide is to allow the expressed protein to be directed from the endoplasmic reticulum to the Golgi apparatus and further to a secretory vesicle for secretion into the culture medium (i.e. exportation of the protein across the cell wall or at least through the cellular membrane into the periplasmic space of the yeast cell).
  • the leader peptide may be the yeast ⁇ -factor leader (the use of which is described in e.g. US 4,546,082, EP 16 201 , EP 123 294, EP 123 544 and EP 163 529).
  • the leader peptide may be a synthetic leader peptide, which is to say a leader peptide not found in nature.
  • Synthetic leader peptides may, for instance, be constructed as described in WO 89/02463 or WO 92/1 1378.
  • the signal peptide may conveniently be derived from a gene encoding an Aspergillus sp. amylase or glucoamylase, a gene encoding a Rhizomucor miehei lipase or protease or a Humicola lanuginosa lipase.
  • the signal peptide is preferably derived from a gene encoding A. oryzae TAKA amylase, A. niger neutral ⁇ -amylase, A. niger acid-stable amylase, or A niger glucoamylase.
  • the signal peptide may conveniently be derived from an insect gene (cf. WO 90/05783), such as the lepidopteran Manduca sexta adipokinetic hormone precursor signal peptide (cf. US 5,023,328).
  • the procedures used to ligate the DNA sequences coding for the present protein, the promoter and optionally the terminator and/or secretory signal sequence, respectively, and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (cf., for instance, Sambrook et al., op.cit).
  • the host cell into which the DNA construct or the recombinant vector of the invention is introduced may be any cell which is capable of producing the present protein and includes bacteria, yeast, fungi and higher eukaryotic cells.
  • Examples of bacterial host cells which, on cultivation, are capable of producing GH are grampositive bacteria such as strains of Bacillus, such as strains of B. subtilis, B. licheniformis, B. lentus, B. brevis, B. stearothermophilus, B. alkalophilus, B. 38
  • amyloliquefaciens B. coagulans, B. circulans, B. lautus, B. megatherium or B. thuringiensis, or strains of Streptomyces, such as S. lividans or S. murinus, or gramnegative bacteria such as Echerichia coli.
  • the transformation of the bacteria may be effected by protoplast transformation or by using competent cells in a manner known per se (cf. Sambrook et al., supra).
  • the protein When expressing a protein in bacteria such as E. coli, the protein may be retained in the cytoplasm, typically as insoluble granules (known as inclusion bodies), or may be directed to the periplasmic space by a bacterial secretion sequence. In the former case, the cells are lysed and the granules are recovered and denatured after which the protein is refolded by diluting the denaturing agent. In the latter case, the protein may be recovered from the periplasmic space by disrupting the cells, e.g. by sonication or osmotic shock, to release the contents of the periplasmic space and recovering the protein.
  • sonication or osmotic shock to release the contents of the periplasmic space and recovering the protein.
  • suitable mammalian cell lines are the COS (ATCC CRL 1650), BHK (ATCC CRL 1632, ATCC CCL 10), CHL (ATCC CCL39) or CHO (ATCC CCL 61 ) cell lines.
  • Methods of transfecting mammalian cells and expressing DNA sequences introduced in the cells are described in e.g. Kaufman and Sharp, J. MoI. Biol. 159 (1982), 601 - 621 ; Southern and Berg, J. MoI. Appl. Genet. 1 (1982), 327 - 341 ; Loyter et al. , Proc. Natl. Acad. Sci.
  • yeasts cells include cells of Saccharomyces spp. or Schizosaccharomyces spp., in particular strains of Saccharomyces cerevisiae or Saccharomyces reteyveri. Methods for transforming yeast cells with heterologous DNA and producing heterologous proteins therefrom are described, e.g. in US 4,599,311 , US 4,931 ,373, US 4,870,008, 5,037,743, and US 4,845,075, all of which are hereby incorporated by reference. Transformed cells are selected by a phenotype determined by a selectable marker, commonly drug resistance or the ability to grow in the absence of a particular nutrient, e.g. leucine.
  • a selectable marker commonly drug resistance or the ability to grow in the absence of a particular nutrient, e.g. leucine.
  • a preferred vector for use in yeast is the POT1 vector disclosed in US 4,931 ,373.
  • the DNA sequence encoding a protein of the invention may be preceded by a signal sequence and optionally a leader sequence , e.g. as described above.
  • suitable yeast cells are strains of Kluyveromyces, such as K. lactis, Hansenula, e.g. H. polymorpha, or Pichia, e.g. P. pastoris (cf. Gleeson et al., J. Gen. Microbiol. 132, 1986, pp. 3459-3465; US 4,882,279).
  • Examples of other fungal cells are cells of filamentous fungi, e.g. Aspergillus spp., Neurospora spp., Fusarium spp. or Trichoderma spp., in particular strains of A. oryzae, A. 39
  • a filamentous fungus When a filamentous fungus is used as the host cell, it may be transformed with the DNA construct of the invention, conveniently by integrating the DNA construct in the host chromosome to obtain a recombinant host cell.
  • This integration is generally considered to be an advantage as the DNA sequence is more likely to be stably maintained in the cell.
  • Integra ⁇ tion of the DNA constructs into the host chromosome may be performed according to conventional methods, e.g. by homologous or heterologous recombination. Transformation of insect cells and production of heterologous proteins therein may be performed as described in US 4,745,051 ; US 4,879,236; US 5,155,037; 5,162,222; EP 397,485) all of which are incorporated herein by reference.
  • the insect cell line used as the host may suitably be a Lepidoptera cell line, such as Spodoptera frugiperda cells or Trichoplusia ni cells (cf. US 5,077,214). Culture conditions may suitably be as described in, for instance, WO 89/01029 or WO 89/01028, or any of the aforementioned references.
  • the transformed or transfected host cell described above is then cultured in a suitable nutrient medium under conditions permitting the expression of the present protein, after which the resulting protein is recovered from the culture.
  • the medium used to culture the cells may be any conventional medium suitable for growing the host cells, such as minimal or complex media containing appropriate supplements. Suitable media are available from commercial suppliers or may be prepared according to published recipes (e.g. in catalogues of the American Type Culture Collection).
  • the protein produced by the cells may then be recovered from the culture medium by conventional procedures including separating the host cells from the medium by centrifugation or filtration, precipitating the proteinaceous components of the supernatant or filtrate by means of a salt, e.g. ammonium sulphate, purification by a variety of chromatographic procedures, e.g. ion exchange chromatography, gelfiltration chromatography, affinity chromatography, or the like, dependent on the specific protein in question.
  • a salt e.g. ammonium sulphate
  • compositions comprising a conjugated GH of the present invention which is present in a concentration from 10 '15 mg/ml to 200 mg/ml, such as e.g. 10 ⁇ 10 mg/ml to 5 mg/ml and wherein said composition has a pH from 2.0 to 10.0.
  • the composition may further comprise a buffer system, preservative(s), 40
  • the pharmaceutical composition is an aqueous composition, i.e. composition comprising water. Such composition is typically a solution or a suspension.
  • the pharmaceutical composition is an aqueous solution.
  • aqueous composition is defined as a composition comprising at least 50 % w/w water.
  • aqueous solution is defined as a solution comprising at least 50 %w/w water
  • aqueous suspension is defined as a suspension comprising at least 50 %w/w water.
  • the pharmaceutical composition is a freeze-dried composition, whereto the physician or the patient adds solvents and/or diluents prior to use.
  • the pharmaceutical composition is a dried composition (e.g. freeze-dried or spray-dried) ready for use without any prior dissolution.
  • the invention in a further aspect relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an aqueous solution of a GH conjugate, and a buffer, wherein said GH conjugate is present in a concentration from 0.1 -100 mg/ml or above, and wherein said composition has a pH from about 2.0 to about 10.0.
  • the pH of the composition is selected from the list consisting of 2.0, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1 , 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1 , 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1 , 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1 , 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1 , 7.2, 7.3, 7.4, 7.5, 7.6, 7.7,
  • the buffer is selected from the group consisting of sodium acetate, sodium carbonate, citrate, glycylglycine, histidine, glycine, lysine, arginine, sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium phosphate, and tris(hydroxymethyl)-aminomethan, bicine, tricine, malic acid, succinate, maleic acid, fumaric acid, tartaric acid, aspartic acid or mixtures thereof.
  • Each one of these specific buffers constitutes an alternative embodiment of the invention.
  • the composition further comprises a pharmaceutically acceptable preservative.
  • the preservative is selected from the group consisting of phenol, o-cresol, m-cresol, p-cresol, methyl p-hydroxybenzoate, propyl p-hydroxybenzoate, 2-phenoxyethanol, butyl p- hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, bronopol, benzoic acid, imidurea, chlorohexidine, sodium dehydroacetate, chlorocresol, ethyl p- hydroxybenzoate, benzethonium chloride, chlorphenesine (3p-chlorphenoxypropane-1 ,2-diol) 41
  • the preservative is present in a concentration from 0.1 mg/ml to 20 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 0.1 mg/ml to 5 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 5 mg/ml to 10 mg/ml. In a further embodiment of the invention the preservative is present in a concentration from 10 mg/ml to 20 mg/ml. Each one of these specific preservatives constitutes an alternative embodiment of the invention.
  • the use of a preservative in pharmaceutical compositions is well-known to the skilled person.
  • the composition further comprises an isotonic agent.
  • the isotonic agent is selected from the group consisting of a salt (e.g. sodium chloride), a sugar or sugar alcohol, an amino acid (e.g. L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine), an alditol (e.g.
  • glycerol glycerine
  • 1 ,2-propanediol propyleneglycol
  • 1 ,3-propanediol 1 ,3- butanediol
  • polyethyleneglycol e.g. PEG400
  • Any sugar such as mono-, di-, or polysaccharides, or water-soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, soluble starch, hydroxyethyl starch and carboxymethylcellulose-Na may be used.
  • the sugar additive is sucrose.
  • Sugar alcohol is defined as a C4-C8 hydrocarbon having at least one -OH group and includes, for example, mannitol, sorbitol, inositol, galactitol, dulcitol, xylitol, and arabitol.
  • the sugar alcohol additive is mannitol.
  • the sugars or sugar alcohols mentioned above may be used individually or in combination. There is no fixed limit to the amount used, as long as the sugar or sugar alcohol is soluble in the liquid preparation and does not adversely effect the stabilizing effects obtained using the methods of the invention.
  • the sugar or sugar alcohol concentration is between about 1 mg/ml and about 150 mg/ml.
  • the isotonic agent is present in a concentration from 1 mg/ml to 50 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 1 mg/ml to 7 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 8 mg/ml to 24 mg/ml. In a further embodiment of the invention the isotonic agent is present in a concentration from 25 mg/ml to 50 mg/ml. Each one of these specific isotonic agents constitutes an alternative embodiment of the invention.
  • the use of an isotonic agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 20 th edition, 2000. 42
  • the composition further comprises a chelating agent.
  • the chelating agent is selected from salts of ethylenediaminetetraacetic acid (EDTA), citric acid, and aspartic acid, and mixtures thereof.
  • the chelating agent is present in a concentration from 0.1 mg/ml to 5mg/ml.
  • the chelating agent is present in a concentration from 0.1 mg/ml to 2mg/ml.
  • the chelating agent is present in a concentration from 2mg/ml to 5mg/ml.
  • Each one of these specific chelating agents constitutes an alternative embodiment of the invention.
  • the use of a chelating agent in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 20 th edition, 2000.
  • composition further comprises a stabilizer.
  • a stabilizer in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington: The Science and Practice of Pharmacy, 20 th edition, 2000.
  • compositions of the invention are stabilized liquid pharmaceutical compositions whose therapeutically active components include a protein that possibly exhibits aggregate formation during storage in liquid pharmaceutical compositions.
  • aggregate formation is intended a physical interaction between the protein molecules that results in formation of oligomers, which may remain soluble, or large visible aggregates that precipitate from the solution.
  • during storage is intended a liquid pharmaceutical composition or composition once prepared, is not immediately administered to a subject. Rather, following preparation, it is packaged for storage, either in a liquid form, in a frozen state, or in a dried form for later reconstitution into a liquid form or other form suitable for administration to a subject.
  • liquid pharmaceutical composition or composition is dried either by freeze drying (i.e., lyophilization; see, for example, Williams and PoIIi (1984) J. Parenteral Sci. Technol. 38:48-59), spray drying (see Masters (1991 ) in Spray-Drying Handbook (5th ed; Longman Scientific and Technical, Essez, U.K.), pp. 491 - 676; Broadhead et al. (1992) Drug Devel. Ind. Pharm. 18:1 169-1206; and Mumenthaler et al. (1994) Pharm. Res. 1 1 :12-20), or air drying (Carpenter and Crowe (1988) Cryobiology
  • membranes or pumps when the protein-containing pharmaceutical composition is administered using an infusion system.
  • compositions of the invention may further comprise an amount of an amino acid base sufficient to decrease aggregate formation by the protein during storage of the composition.
  • amino acid base is intended an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. Where a combination of amino acids is used, all of the amino acids may be present in their free base forms, all may be present in their salt forms, or some may be present in their free base forms while others are present in their salt forms.
  • amino acids to use in preparing the compositions of the invention are those carrying a charged side chain, such as arginine, lysine, aspartic acid, and glutamic acid.
  • Any stereoisomer i.e., L or D isomer, or mixtures thereof
  • a particular amino acid methionine, histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof
  • an organic base such as but not limited to imidazole
  • the L-stereoisomer of an amino acid is used.
  • the L- stereoisomer is used.
  • Compositions of the invention may also be formulated with analogues of these amino acids.
  • amino acid analogue is intended a derivative of the naturally occurring amino acid that brings about the desired effect of decreasing aggregate formation by the protein during storage of the liquid pharmaceutical compositions of the invention.
  • Suitable arginine analogues include, for example, aminoguanidine, ornithine and N- monoethyl L-arginine
  • suitable methionine analogues include ethionine and buthionine
  • suitable cysteine analogues include S-methyl-L cysteine.
  • the amino acid analogues are incorporated into the compositions in either their free base form or their salt form.
  • the amino acids or amino acid analogues are used in a concentration, which is sufficient to prevent or delay aggregation of the protein.
  • methionine (or other sulphuric amino acids or amino acid analogous) may be added to inhibit oxidation of methionine residues to methionine sulfoxide when the protein acting as the therapeutic agent is a protein comprising at least one methionine residue susceptible to such oxidation.
  • inhibitor is intended minimal accumulation of methionine oxidized species over time. Inhibiting methionine oxidation results in greater retention of the protein in its proper molecular form. Any stereoisomer of methionine (L or D isomer) or any combinations thereof can be used. The amount to be 44
  • the composition contains no more than about 10% to about 30% methionine sulfoxide.
  • this can be obtained by adding methionine such that the ratio of methionine added to methionine residues ranges from about 1 :1 to about 1000:1 , such as 10:1 to about 100:1 .
  • the composition further comprises a stabilizer selected from the group of high molecular weight polymers or low molecular compounds.
  • the stabilizer is selected from polyethylene glycol (e.g. PEG 3350), polyvinyl alcohol (PVA), polyvinylpyrrolidone, carboxy- /hydroxycellulose or derivates thereof (e.g. HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, sulphur-containing substances as monothioglycerol, thioglycolic acid and 2- methylthioethanol, and different salts (e.g. sodium chloride).
  • PEG 3350 polyethylene glycol
  • PVA polyvinyl alcohol
  • PVC polyvinylpyrrolidone
  • carboxy- /hydroxycellulose or derivates thereof e.g. HPC, HPC-SL, HPC-L and HPMC
  • cyclodextrins e.g. sulphur-containing substances as monothioglycerol, thi
  • compositions may also comprise additional stabilizing agents, which further enhance stability of a therapeutically active protein therein.
  • Stabilizing agents of particular interest to the present invention include, but are not limited to, methionine and EDTA, which protect the protein against methionine oxidation, and a nonionic surfactant, which protects the protein against aggregation associated with freeze-thawing or mechanical shearing.
  • the composition further comprises a surfactant.
  • the surfactant is selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, polyoxypropylene-polyoxyethylene block polymers (eg. poloxamers such as Pluronic ® F68, poloxamer 188 and 407, Triton X-100 ), polyoxyethylene sorbitan fatty acid esters, polyoxyethylene and polyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g.
  • Tween-20, Tween-40, Tween-80 and Brij-35 monoglycerides or ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, alcohols, glycerol, lectins and phospholipids (eg. phosphatidyl serine, phosphatidyl choline, phosphatidyl ethanolamine, phosphatidyl inositol, diphosphatidyl glycerol and sphingomyelin), derivates of phospholipids (eg. dipalmitoyl phosphatidic acid) and lysophospholipids (eg.
  • phospholipids eg. dipalmitoyl phosphatidic acid
  • lysophospholipids eg.
  • polar head group that is cholines, ethanolamines, phosphatidic acid, serines, threonines, glycerol, inositol, and the positively charged DODAC, DOTMA, DCP, BISHOP, lysophosphatidylserine and lysophosphatidylthreonine, and glycerophospholipids (eg. cephalins), glyceroglycolipids (eg. galactopyransoide), sphingoglycolipids (eg. ceramides, gangliosides), dodecylphosphocholine, hen egg lysolecithin, fusidic acid derivatives- (e.g.
  • N-alkyl-N,N-dimethylammonio-1 -propanesulfonates 3-cholamido-1 -propyldimethylammonio-1 -propanesulfonate
  • cationic surfactants quaternary ammonium bases
  • cetyl-trimethylammonium bromide cetylpyridinium chloride
  • non- ionic surfactants eg. Dodecyl ⁇ -D-glucopyranoside
  • poloxamines eg.
  • Tetronic's which are tetrafunctional block copolymers derived from sequential addition of propylene oxide and ethylene oxide to ethylenediamine, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. Each one of these specific surfactants constitutes an alternative embodiment of the invention.
  • Such additional ingredients may include wetting agents, emulsifiers, antioxidants, bulking agents, tonicity modifiers, chelating agents, metal ions, oleaginous vehicles, proteins (e.g., human serum albumin, gelatine or proteins) and a zwitterion (e.g., an amino acid such as betaine, taurine, arginine, glycine, lysine and histidine).
  • additional ingredients should not adversely affect the overall stability of the pharmaceutical composition of the present invention.
  • compositions containing a GH conjugate according to the present invention may be administered to a patient in need of such treatment at several sites, for example, at topical sites, for example, skin and mucosal sites, at sites which bypass absorption, for example, administration in an artery, in a vein, in the heart, and at sites which involve absorption, for example, administration in the skin, under the skin, in a muscle or in the abdomen.
  • topical sites for example, skin and mucosal sites
  • sites which bypass absorption for example, administration in an artery, in a vein, in the heart
  • sites which involve absorption for example, administration in the skin, under the skin, in a muscle or in the abdomen.
  • Administration of pharmaceutical compositions according to the invention may be through several routes of administration, for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment.
  • routes of administration for example, lingual, sublingual, buccal, in the mouth, oral, in the stomach and intestine, nasal, pulmonary, for example, through the bronchioles and alveoli or a combination thereof, epidermal, dermal, transdermal, vaginal, rectal, ocular, for examples through the conjunctiva, uretal, and parenteral to patients in need of such a treatment.
  • compositions of the current invention may be administered in several dosage forms, for example, as solutions, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinses, capsules, for example, hard gelatine capsules and soft gelatine capsules, suppositories, rectal capsules, drops, gels, sprays, powder, aerosols, inhalants, eye drops, ophthalmic ointments, ophthalmic rinses, vaginal pessaries, vaginal rings, vaginal ointments, injection solution, in situ transforming solutions, for example in situ gelling, in situ setting, in situ precipitating, in situ crystallization, infusion solution, and implants.
  • solutions for example, suspensions, emulsions, microemulsions, multiple emulsion, foams, salves, pastes, plasters, ointments, tablets, coated tablets, rinses,
  • compositions of the invention may further be compounded in, or attached to, for example through covalent, hydrophobic and electrostatic interactions, a drug carrier, drug delivery system and advanced drug delivery system in order to further enhance stability of the GH conjugate, increase bioavailability, increase solubility, decrease adverse effects, achieve chronotherapy well known to those skilled in the art, and increase patient compliance or any combination thereof.
  • carriers, drug delivery systems and advanced drug delivery systems include, but are not limited to, polymers, for example cellulose and derivatives, polysaccharides, for example dextran and derivatives, starch and derivatives, polyvinyl alcohol), acrylate and methacrylate polymers, polylactic and polyglycolic acid and block co-polymers thereof, polyethylene glycols, carrier proteins, for example albumin, gels, for example, thermogelling systems, for example block co-polymeric systems well known to those skilled in the art, micelles, liposomes, microspheres, nanoparticulates, liquid crystals and dispersions thereof, L2 phase and dispersions there of, well known to those skilled in the art of phase behaviour in lipid-water systems, polymeric 47
  • micelles multiple emulsions, self-emulsifying, self-microemulsifying, cyclodextrins and derivatives thereof, and dendrimers.
  • compositions of the current invention are useful in the composition of solids, semisolids, powder and solutions for pulmonary administration of GH conjugate, using, for example a metered dose inhaler, dry powder inhaler and a nebulizer, all being devices well known to those skilled in the art.
  • compositions of the current invention are specifically useful in the composition of controlled, sustained, protracting, retarded, and slow release drug delivery systems. More specifically, but not limited to, compositions are useful in composition of parenteral controlled release and sustained release systems (both systems leading to a many-fold reduction in number of administrations), well known to those skilled in the art. Even more preferably, are controlled release and sustained release systems administered subcutaneous.
  • examples of useful controlled release system and compositions are hydrogels, oleaginous gels, liquid crystals, polymeric micelles, microspheres, nanoparticles,
  • Methods to produce controlled release systems useful for compositions of the current invention include, but are not limited to, crystallization, condensation, co- crystallization, precipitation, co-precipitation, emulsification, dispersion, high pressure homogenisation, encapsulation, spray drying, microencapsulating, coacervation, phase separation, solvent evaporation to produce microspheres, extrusion and supercritical fluid processes.
  • General reference is made to Handbook of Pharmaceutical Controlled Release (Wise, D.L., ed. Marcel Dekker, New York, 2000) and Drug and the Pharmaceutical Sciences vol. 99: Protein Composition and Delivery (MacNally, E. J., ed. Marcel Dekker, New York, 2000).
  • Parenteral administration may be performed by subcutaneous, intramuscular, intraperitoneal or intravenous injection by means of a syringe, optionally a pen-like syringe.
  • parenteral administration can be performed by means of an infusion pump.
  • a further option is a composition which may be a solution or suspension for the administration of the GH conjugate in the form of a nasal or pulmonal spray.
  • the pharmaceutical compositions containing the GH conjugate of the invention can also be adapted to transdermal administration, e.g. by needle-free injection or from a patch, optionally an iontophoretic patch, or transmucosal, e.g. buccal, administration.
  • stabilized composition refers to a composition with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • physical stability of the protein composition refers to the tendency of the protein to form biologically inactive and/or insoluble aggregates of the protein as a result of exposure of the protein to thermo-mechanical stresses and/or interaction with interfaces and surfaces that are destabilizing, such as hydrophobic surfaces and interfaces.
  • Physical stability of the aqueous protein compositions is evaluated by means of visual inspection and/or turbidity measurements after exposing the composition filled in suitable containers (e.g. cartridges or vials) to mechanical/physical stress (e.g. agitation) at different temperatures for various time periods. Visual inspection of the compositions is performed in a sharp focused light with a dark background.
  • the turbidity of the composition is characterized by a visual score ranking the degree of turbidity for instance on a scale from 0 to 3 (a composition showing no turbidity corresponds to a visual score 0, and a composition showing visual turbidity in daylight corresponds to visual score 3).
  • a composition is classified physical unstable with respect to protein aggregation, when it shows visual turbidity in daylight.
  • the turbidity of the composition can be evaluated by simple turbidity measurements well-known to the skilled person.
  • Physical stability of the aqueous protein compositions can also be evaluated by using a spectroscopic agent or probe of the conformational status of the protein.
  • the probe is preferably a small molecule that preferentially binds to a non-native conformer of the protein.
  • Thioflavin T is a fluorescent dye that has been widely used for the detection of amyloid fibrils. In the presence of fibrils, and perhaps other protein configurations as well, Thioflavin T gives rise to a new excitation maximum at about 450 nm and enhanced emission at about 482 nm when bound to a fibril protein form. Unbound Thioflavin T is essentially non-fluorescent at the wavelengths.
  • Other small molecules can be used as probes of the changes in protein structure from native to non-native states. For instance the "hydrophobic patch" probes that bind preferentially to exposed hydrophobic patches of a protein.
  • the hydrophobic patches are generally buried within the tertiary structure of a protein in its native state, but become exposed as a protein begins to unfold or denature.
  • these small molecular, spectroscopic probes are aromatic, hydrophobic dyes, such as antrhacene, acridine, phenanthroline or the like.
  • Other spectroscopic probes are metal-amino acid complexes, such as cobalt metal complexes of hydrophobic amino acids, such as phenylalanine, leucine, isoleucine, methionine, and valine, or the like.
  • chemical stability of the protein composition as used herein refers to chemical covalent changes in the protein structure leading to formation of chemical 49
  • degradation products with potential less biological potency and/or potential increased immunogenic properties compared to the native protein structure.
  • Various chemical degradation products can be formed depending on the type and nature of the native protein and the environment to which the protein is exposed. Elimination of chemical degradation can most probably not be completely avoided and increasing amounts of chemical degradation products is often seen during storage and use of the protein composition as well-known by the person skilled in the art.
  • Most proteins are prone to deamidation, a process in which the side chain amide group in glutaminyl or asparaginyl residues is hydrolysed to form a free carboxylic acid.
  • a “stabilized composition” refers to a composition with increased physical stability, increased chemical stability or increased physical and chemical stability.
  • a composition must be stable during use and storage (in compliance with recommended use and storage conditions) until the expiration date is reached.
  • the pharmaceutical composition comprising the GH conjugate is stable for more than 6 weeks of usage and for more than 3 years of storage. In another embodiment of the invention the pharmaceutical composition comprising the GH conjugate is stable for more than 4 weeks of usage and for more than 3 years of storage.
  • the pharmaceutical composition comprising the GH conjugateis stable for more than 4 weeks of usage and for more than two years of storage. 50
  • the pharmaceutical composition comprising the GH conjugate is stable for more than 2 weeks of usage and for more than two years of storage.
  • HOAt 3-hydroxy-3H-[1 ,2,3]triazolo[4,5-b]pyridine, 4-aza-3- hydroxybenzotriazole
  • HOBt N-hydroxybenzotriazole, 1 -hydroxybenzotriazole
  • HONSu N-hydroxysuccinimide
  • NMP N-methylpyrrolidone
  • HPLC high pressure liquid chromatography 52
  • HPLC-systems from Merck-Hitachi HibarTM RT 250-4, LichrosorbTM RP 18, 5.0 ⁇ m, 4.0 x 250 mm, gradient elution, 20% to 80% acetonitrile in water within 30 min, 1 .0 ml/min, detection at 254 nm
  • Waters SymmetryTM, C18, 3.5 ⁇ m, 3.0 x 150 mm, gradient elution, 5% to 90% acetonitrile in water within 15 min, 1 .0 ml/min, detection at 214 nm
  • condensation reaction to yield compounds of formula (I) or (T) may also be performed directly by mixing compounds of formula (II), in which Q is an acetal-type structure, with a suitable alkoxylamine, hydrazine, or aminothiol, in the presence of an acid or a silver salt or a mercury salt, as described in the literature (Tumelty et al., J. Am. Chem. Soc. 2003, 125, 14238-14239).
  • the compounds of formula (V) according to the invention may be prepared as follows:
  • Condensation of compounds of general formula (III) or of general formula (V) with alkoxylamines, hydrazines, or aminothiols is performed by adding a solution of a suitable alkoxylamine, hydrazine, or aminothiol in water or in a mixture of water and a dipolar solvent or in a dipolar solvent alone, such as DMF, to the solution of compounds (III) or (V).
  • the pH of the resulting mixture may range from 2.0 to 12.0, and may optionally be adjusted to a given value, for instance 3.6, 4.0, 6.0, 6.5, 10.0 or 10.5.
  • the resulting mixture is shaken carefully for 1 -200 h, and the reaction is followed by HPLC or any other, conventional analytical tool. When the reaction is finished the mixture is diluted with water, and the product purified using standard chromatographic techniques.
  • Ser-hGH analogue expression plasmid was created on the basis of pNNC13 (Zbasic2mt-D4K-hGH), which expresses the wild type hGH in fusion with Zbasic domain
  • E. coli BL21 (DE3) was transformed by pET1 1 a-Zbasic2mt-D4K-Ser-hGH. Single colony was inoculated into 100ml LB media with 100 ⁇ g/ml Amp and grew at 37 0 C. When OD600 reached 0.6, the cell culture temperature was reduced to 3O 0 C, and the cells were induced with 1 mM IPTG for 4 hours at 30 degree. The bacteria cells were harvested by centrifugation at 300Og for 15 minutes (Eppendorf centrifuge 5810R).
  • the cell pellet was re- suspended in cell lysis buffer (25 mM Na 2 HPO 4 25 mM NaH 2 PO 4 pH 7, 5 mM EDTA, 0.1 % Triton X-100), and the cells were disrupted by cell disruption at 30 kpsi (Constant Cell Disruption Systems).
  • the lysate was clarified by centrifugation at 1000Og for 30 minutes. The supernatant was saved and used for purification, while the pellet was discarded.
  • Zbasic2mt-D4K-Ser-hGH was purified on SP-Sepharose using a step gradient elution (buffer A: 25 mM Na 2 HPO 4 25 mM NaH 2 PO 4 pH 7; buffer B: 25 mM Na 2 HPO 4 25 mM NaH 2 PO 4 pH 7, 1 M NaCI). The protein was subsequently cleaved using Enteropeptidase for the release of Ser-hGH.
  • Ser-hGH was further purified on a Butyl Sepharose 4FF column to separate the product from the Zbasic2mt-D4K domain and Enteropeptidase (buffer A: 100 mM Hepes pH 7.5; buffer B: 100 mM Hepes pH 7.5, 2 M NaCI, a linear gradient was used).
  • buffer A 100 mM Hepes pH 7.5
  • buffer B 100 mM Hepes pH 7.5, 2 M NaCI, a linear gradient was used.
  • the final product of Ser-hGH was buffer exchanged and lyophilized from 50 mM NH 4 HCO 3 , pH 7.8.
  • Ser-hGH (Mwt: 2221 1 ; approx 1 mg, 45 nmol) was dissolved in the buffer (110 ⁇ l). To this solution was added the methionine solution (10 ⁇ l, approx 30 eq) followed by the NaIO 4 solution (10 ⁇ l, approx 5 eq). The clear solution was kept at room temperature for 15 min. A solution of the hydroxylamine (10 ⁇ l; approx 10 eq) was added and the resulting, slightly turbid mixture was kept at room temperature for 15 min.
  • Example 3 1 -[2-(O-(4-(4-(Methoxy-PEG(30K)-oxy)butyrylamino)butyl)oximino)acetyl]- hGH
  • Buffer A triethanolamine (270 mg, 1 .8 mmol), 3-methylthiopropanol (580 mg, 5.46 mmol), water (40 ml).
  • Buffer F L-methionine (2.0 g) + water (80 ml).
  • buffer F (Amicon Ultra, cut-off 5000 Da, 10 0 C). The residue was diluted with buffer F to 1.5 ml and to this solution ice-cold (0 0 C) NMP (0.5 ml) was added.
  • BAF-3GHR assay to determine growth hormone activity The BAF-3 cells (a murine pro-B lymphoid cell line derived from the bone marrow) was originally IL-3 dependent for growth and survival. II-3 activates JAK-2 and STAT which are the same mediators GH is activating upon stimulation. After transfection of the human growth hormone receptor the cell line was turn into a growth hormone-dependent cell line. This clone can be used to evaluate the effect of different growth hormone samples on the survival of the BAF-3GHR.
  • the BAF-3GHR cells are grown in starvation medium (culture medium without growth hormoen) for 24 hours at 37 0 C, 5 % CO 2 .
  • the cells are washed and re-suspended in starvation medium and seeded in plates. 10 ⁇ l of growth hormone compound or human growth hormone in different concentrations or control is added to the cells, and the plates are incubated for 68 hours at 37 0 C, 5 % CO 2 .
  • AlamarBlue® is added to each well and the cells are then incubated for another 4 hours.
  • the AlamarBlue® is a redox indicator, and is reduced by reactions innate to cellular metabolism and, therefore, provides an indirect measure of viable cell number. Finally, the metabolic activity of the cells is measure in a fluorescence plate reader.
  • the absorbance in the samples is expressed in % of cells not stimulated with growth hormone compound or control and from the concentration-response curves the activity (amount of a compound that stimulates the cells with 50%) can be calculated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Endocrinology (AREA)
  • Biomedical Technology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Reproductive Health (AREA)
  • Toxicology (AREA)
  • Psychiatry (AREA)
  • Pain & Pain Management (AREA)
  • Gynecology & Obstetrics (AREA)
  • Obesity (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Tropical Medicine & Parasitology (AREA)

Abstract

L'invention concerne des composés de formule (I), dans laquelle GH représente un radical dérivé d'un composé d'hormone de croissance par retrait d'un atome d'hydrogène du groupe amino N-terminal ; X représente oxygène ou deux atomes d'hydrogène ; Z représente une liaison, alkylène, arylène, hétéroarylène, -CH2-O-(CH2)1-10-, -CH2-O- (C6H4)- ou des combinaisons de ces derniers et Y représente un radical sélectionné parmi (II), (III), (IV) et (V). Cette invention concerne également des procédés pour produire lesdits composés, lesquels sont utiles en thérapie.
PCT/EP2005/055336 2004-10-18 2005-10-18 Conjugues d'hormones de croissance WO2006042848A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2007537265A JP2008516621A (ja) 2004-10-18 2005-10-18 成長ホルモン結合体
US11/577,238 US20080182783A1 (en) 2004-10-18 2005-10-18 Growth Hormone Conjugates
EP05797317A EP1805216A2 (fr) 2004-10-18 2005-10-18 Conjugues d'hormones de croissance

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DKPA200401588 2004-10-18
DKPA200401588 2004-10-18
DKPA200500107 2005-01-21
DKPA200500107 2005-01-21

Publications (3)

Publication Number Publication Date
WO2006042848A2 true WO2006042848A2 (fr) 2006-04-27
WO2006042848A3 WO2006042848A3 (fr) 2006-09-21
WO2006042848A9 WO2006042848A9 (fr) 2007-05-18

Family

ID=36203319

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/055336 WO2006042848A2 (fr) 2004-10-18 2005-10-18 Conjugues d'hormones de croissance

Country Status (4)

Country Link
US (1) US20080182783A1 (fr)
EP (1) EP1805216A2 (fr)
JP (1) JP2008516621A (fr)
WO (1) WO2006042848A2 (fr)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007025988A2 (fr) * 2005-08-30 2007-03-08 Novo Nordisk Health Care Ag Formulations liquides d'hormone de croissance pegylee
WO2009065126A2 (fr) * 2007-11-16 2009-05-22 Boston Protein Solutions Excipients pour la stabilisation de protéines
JP2010505875A (ja) * 2006-10-04 2010-02-25 ノヴォ ノルディスク アー/エス グリセロール連結のpeg化された糖および糖ペプチド
WO2010029107A1 (fr) * 2008-09-09 2010-03-18 Novo Nordisk Health Care Ag Conjugué d’hormone de croissance doté d’une stabilité accrue
EP2727605A2 (fr) * 2011-06-28 2014-05-07 B&L Delipharm, Corp. Analogue d'exendine-4 pégylé par du polyéthylène glycol ou un dérivé de celui-ci, son procédé de préparation et composition pharmaceutique pour prévenir ou traiter le diabète, le contenant en tant que principe actif
US9175061B2 (en) 2006-07-07 2015-11-03 Novo Nordisk Health Care Ag Protein conjugates and methods for their preparation
US9186323B2 (en) 2007-05-02 2015-11-17 Novo Nordisk Healthcare Ag High concentration factor VII polypeptide formulations comprising an aromatic preservative and an antioxidant
US9321825B2 (en) 2006-06-12 2016-04-26 Theraly Pharmaceuticals Inc. N-terminal modified PEG-TRAIL
US10988531B2 (en) 2014-09-03 2021-04-27 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
US11007251B2 (en) 2015-12-17 2021-05-18 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
US11084879B2 (en) 2016-04-07 2021-08-10 The Johns Hopkins University Compositions and methods for treating pancreatitis and pain with death receptor agonists
US11123405B2 (en) 2015-12-23 2021-09-21 The Johns Hopkins University Long-acting GLP-1R agonist as a therapy of neurological and neurodegenerative conditions

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993000109A1 (fr) * 1991-06-28 1993-01-07 Genentech, Inc. Procede de stimulation de la reponse immunitaire a l'aide d'hormone de croissance
WO1997011178A1 (fr) * 1995-09-21 1997-03-27 Genentech, Inc. Variants de l'hormone de croissance humaine
WO2004007687A2 (fr) * 2002-07-16 2004-01-22 Stuart Bussell Methodes de modelisation d'adn multimere et de sequences de proteines polymeres par fusions directes ou a l'aide de lieurs
US20040127417A1 (en) * 2002-11-20 2004-07-01 Finn Rory F. N-terminally monopegylated human growth hormone conjugates and process for their preparation

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69231467T2 (de) * 1991-05-10 2001-01-25 Genentech Inc Auswählen von agonisten und antagonisten von liganden
WO1999045026A1 (fr) * 1998-03-05 1999-09-10 Chiron Corporation Procede d'augmentation de la demi-vie serique d'une molecule biologiquement active

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993000109A1 (fr) * 1991-06-28 1993-01-07 Genentech, Inc. Procede de stimulation de la reponse immunitaire a l'aide d'hormone de croissance
WO1997011178A1 (fr) * 1995-09-21 1997-03-27 Genentech, Inc. Variants de l'hormone de croissance humaine
WO2004007687A2 (fr) * 2002-07-16 2004-01-22 Stuart Bussell Methodes de modelisation d'adn multimere et de sequences de proteines polymeres par fusions directes ou a l'aide de lieurs
US20040127417A1 (en) * 2002-11-20 2004-07-01 Finn Rory F. N-terminally monopegylated human growth hormone conjugates and process for their preparation

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CLARK R ET AL: "Long-acting growth hormones produced by conjugation with polyethylene glycol" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOCHEMICAL BIOLOGISTS, BIRMINGHAM,, US, vol. 271, no. 36, 6 September 1996 (1996-09-06), pages 21969-21977, XP002216386 ISSN: 0021-9258 *
GAERTNER HUBERT F ET AL: "Site-specific attachment of functionalized poly(ethylene glycol) to the amino terminus of proteins" BIOCONJUGATE CHEMISTRY, vol. 7, no. 1, 1996, pages 38-44, XP002384087 ISSN: 1043-1802 *
GEOGHEGAN K F: "Site-directed conjugation of nonpeptide groups to peptides and proteins via periodate oxidation of a 2-amino alcohol. Application to modification at N-terminal serine" BIOCONJUGATE CHEMISTRY, ACS, WASHINGTON, DC, US, vol. 3, no. 2, 1992, pages 138-146, XP002133027 ISSN: 1043-1802 *
See also references of EP1805216A2 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007025988A3 (fr) * 2005-08-30 2007-05-03 Novo Nordisk As Formulations liquides d'hormone de croissance pegylee
WO2007025988A2 (fr) * 2005-08-30 2007-03-08 Novo Nordisk Health Care Ag Formulations liquides d'hormone de croissance pegylee
US8293708B2 (en) 2005-08-30 2012-10-23 Novo Nordisk Health Care A/G Liquid formulations N-terminal serine of pegylated growth hormone
US10046059B2 (en) 2006-06-12 2018-08-14 D&D Pharmatech Inc. Methods of administering an N-terminal modified PEG-TRAIL
US9321825B2 (en) 2006-06-12 2016-04-26 Theraly Pharmaceuticals Inc. N-terminal modified PEG-TRAIL
US9175061B2 (en) 2006-07-07 2015-11-03 Novo Nordisk Health Care Ag Protein conjugates and methods for their preparation
JP2010505875A (ja) * 2006-10-04 2010-02-25 ノヴォ ノルディスク アー/エス グリセロール連結のpeg化された糖および糖ペプチド
US9186323B2 (en) 2007-05-02 2015-11-17 Novo Nordisk Healthcare Ag High concentration factor VII polypeptide formulations comprising an aromatic preservative and an antioxidant
WO2009065126A2 (fr) * 2007-11-16 2009-05-22 Boston Protein Solutions Excipients pour la stabilisation de protéines
WO2009065126A3 (fr) * 2007-11-16 2009-08-06 Boston Protein Solutions Excipients pour la stabilisation de protéines
US8642532B2 (en) 2007-11-16 2014-02-04 Guohan Yang Excipients for protein stabilization
WO2010029107A1 (fr) * 2008-09-09 2010-03-18 Novo Nordisk Health Care Ag Conjugué d’hormone de croissance doté d’une stabilité accrue
EP2727605A4 (fr) * 2011-06-28 2014-12-24 Theraly Pharmaceuticals Inc Analogue d'exendine-4 pégylé par du polyéthylène glycol ou un dérivé de celui-ci, son procédé de préparation et composition pharmaceutique pour prévenir ou traiter le diabète, le contenant en tant que principe actif
EP2727605A2 (fr) * 2011-06-28 2014-05-07 B&L Delipharm, Corp. Analogue d'exendine-4 pégylé par du polyéthylène glycol ou un dérivé de celui-ci, son procédé de préparation et composition pharmaceutique pour prévenir ou traiter le diabète, le contenant en tant que principe actif
US10406230B2 (en) 2011-06-28 2019-09-10 D&D Pharmatech Inc. Exendin-4 analogue pegylated with polyethylene glycol or derivative thereof, preparation method thereof, and pharmaceutical composition for preventing or treating diabetes, containing same as active ingredient
US10988531B2 (en) 2014-09-03 2021-04-27 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
US11732038B2 (en) 2014-09-03 2023-08-22 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
US11007251B2 (en) 2015-12-17 2021-05-18 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
US11123405B2 (en) 2015-12-23 2021-09-21 The Johns Hopkins University Long-acting GLP-1R agonist as a therapy of neurological and neurodegenerative conditions
US11084879B2 (en) 2016-04-07 2021-08-10 The Johns Hopkins University Compositions and methods for treating pancreatitis and pain with death receptor agonists

Also Published As

Publication number Publication date
US20080182783A1 (en) 2008-07-31
WO2006042848A9 (fr) 2007-05-18
WO2006042848A3 (fr) 2006-09-21
JP2008516621A (ja) 2008-05-22
EP1805216A2 (fr) 2007-07-11

Similar Documents

Publication Publication Date Title
US9175061B2 (en) Protein conjugates and methods for their preparation
US20080182783A1 (en) Growth Hormone Conjugates
US20190203213A1 (en) Growth Hormones with Prolonged In-Vivo Efficacy
EP2461831B1 (fr) Hormones de croissance ayant une efficacité in vivo prolongée
US20120088716A1 (en) Polypeptide Protracting Tags
US20100197573A1 (en) Transglutaminase Mediated Conjugation of Growth Hormone
US20090105134A1 (en) C-Terminally Pegylated Growth Hormones
US20080274075A1 (en) Poly(Ethylene Glycol) Derivatives and Process For Their Coupling to Proteins
WO2006018429A1 (fr) Mutlimeres de peptides
ES2745484T3 (es) Hormonas de crecimiento con eficacia prolongada in vivo

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2005797317

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007537265

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWP Wipo information: published in national office

Ref document number: 2005797317

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 11577238

Country of ref document: US